Matches in SemOpenAlex for { <https://semopenalex.org/work/W2073293485> ?p ?o ?g. }
- W2073293485 endingPage "830" @default.
- W2073293485 startingPage "817" @default.
- W2073293485 abstract "The potential for the use of stem/progenitor cells for the restoration of injured or diseased tissues has garnered much interest recently, establishing a new field of research called regenerative medicine. Attention has been focused on embryonic stem cells derived from human fetal tissues. However, the use of human fetal tissue for research and transplantation is controversial. An alternative is the isolation and utilization of multipotent stem/progenitor cells derived from adult donor tissues. We have previously reported on the isolation, propagation, and partial characterization of a population of stem/progenitor cells isolated from the pancreatic islets of Langerhans of adult human donor pancreata. Here we show that these human adult tissue-derived cells, nestin-positive islet-derived stem/progenitor cells, prepared from human adult pancreata survive engraftment and produce tissue chimerism when transplanted into immunocompetent mice either under the kidney capsule or by systemic injection. These xenografts seem to induce immune tolerance by establishing a mixed chimerism in the mice. We propose that a population of stem/progenitor cells isolated from the islets of the pancreas can cross xenogeneic transplantation immune barriers, induce tissue tolerance, and grow. The potential for the use of stem/progenitor cells for the restoration of injured or diseased tissues has garnered much interest recently, establishing a new field of research called regenerative medicine. Attention has been focused on embryonic stem cells derived from human fetal tissues. However, the use of human fetal tissue for research and transplantation is controversial. An alternative is the isolation and utilization of multipotent stem/progenitor cells derived from adult donor tissues. We have previously reported on the isolation, propagation, and partial characterization of a population of stem/progenitor cells isolated from the pancreatic islets of Langerhans of adult human donor pancreata. Here we show that these human adult tissue-derived cells, nestin-positive islet-derived stem/progenitor cells, prepared from human adult pancreata survive engraftment and produce tissue chimerism when transplanted into immunocompetent mice either under the kidney capsule or by systemic injection. These xenografts seem to induce immune tolerance by establishing a mixed chimerism in the mice. We propose that a population of stem/progenitor cells isolated from the islets of the pancreas can cross xenogeneic transplantation immune barriers, induce tissue tolerance, and grow. A revelation in the field of tissue regeneration has been the finding that pluripotent stem cells, or multipotent progenitor cells, exist not only in embryo blastocysts and fetal gonadal ridges but also exist in probably all organs of the adult body.1Blau HM Brazelton TR Weimann JM The evolving concept of a stem cell: entity or function?.Cell. 2001; 105: 829-841Abstract Full Text Full Text PDF PubMed Scopus (934) Google Scholar, 2Clarke DL Johansson CB Wilbertz J Veress B Nilsson E Karlstrom H Lendahl U Frisen J Generalized potential of adult neural stem cells.Science. 2000; 288: 1660-1663Crossref PubMed Scopus (917) Google Scholar, 3Gage FH Mammalian neural stem cells.Science. 2000; 287: 1433-1438Crossref PubMed Scopus (4058) Google Scholar, 4Jiang Y Jahagirdar BN Reinhardt RL Schwartz RE Keene CD Ortiz-Gonzalez XR Reyes M Lenvik T Lund T Blackstad M Du J Aldrich S Lisberg A Low WC Largaespada DA Verfaillie CM Pluripotency of mesenchymal stem cells derived from adult marrow.Nature. 2002; 418: 41-49Crossref PubMed Scopus (5142) Google Scholar, 5Slack JM Stem cells in epithelial tissues.Science. 2000; 287: 1431-1433Crossref PubMed Scopus (317) Google Scholar, 6Van der Kooy D Weiss S Why stem cells?.Science. 2000; 287: 1439-1441Crossref PubMed Scopus (243) Google Scholar, 7Watt FM Hogan BL Out of Eden: stem cells and their niches.Science. 2000; 287: 1427-1430Crossref PubMed Scopus (1470) Google Scholar, 8Weissman IL Anderson DJ Gage F Stem and progenitor cells: origins, phenotypes, lineage commitments, and transdifferentiations.Annu Rev Cell Dev Biol. 2001; 17: 387-403Crossref PubMed Scopus (758) Google Scholar Stem cells manifest at least four important properties: they are highly mobile, have the ability for self renewal, differentiate into different cell lineages given exposure to appropriate local environmental stimuli known as growth factors or morphogens,1Blau HM Brazelton TR Weimann JM The evolving concept of a stem cell: entity or function?.Cell. 2001; 105: 829-841Abstract Full Text Full Text PDF PubMed Scopus (934) Google Scholar and may induce immune tolerance.9Shizuru JA Weissman IL Kernoff R Masek M Sheffold YC Purified hematopoietic stem cell grafts induce tolerance to alloantigens and can mediate positive and negative T cell selection.Proc Natl Acad Sci USA. 2000; 97: 9555-9560Crossref PubMed Scopus (83) Google Scholar Morphogens are typically provided by localized spatial regions of mesenchyme, so called mesenchymal niches. Many studies have been reported about how neural stem cells can be converted into blood (although controversial),10Bjornson CR Rietze RL Reynolds BA Magli MC Vescovi AL Turning brain into blood: a hematopoietic fate adopted by adult neural stem cells in vivo.Science. 1999; 283: 534-537Crossref PubMed Scopus (1288) Google Scholar and hematopoietic stem cells can be converted into brain,11Eglitis MA Mezey E Hematopoietic cells differentiate into both microglia and macroglia in the brains of adult mice.Proc Natl Acad Sci USA. 1997; 94: 4080-4085Crossref PubMed Scopus (949) Google Scholar, 12Zhang ZG Zhang L Jiang Q Chopp M Bone marrow-derived endothelial progenitor cells participate in cerebral neovascularization after focal cerebral ischemia in the adult mouse.Circ Res. 2002; 90: 284-288Crossref PubMed Scopus (417) Google Scholar muscle,13Gussoni E Soneoka Y Strickland CD Buzney EA Khan MK Flint AF Kunkel LM Mulligan RC Dystrophin expression in the mdx mouse restored by stem cell transplantation.Nature. 1999; 401: 390-394Crossref PubMed Scopus (1635) Google Scholar heart,14Jackson KA Majka SM Wang H Pocius J Hartley CJ Majesky MW Entman ML Michael LH Hirschi KK Goodell MA Regeneration of ischemic cardiac muscle and vascular endothelium by adult stem cells.J Clin Invest. 2001; 107: 1395-1402Crossref PubMed Scopus (1766) Google Scholar and liver.15Lagasse E Connors H Al-Dhalimy M Reitsma M Dohse M Osborne L Wang X Finegold M Weissman IL Grompe M Purified hematopoietic stem cells can differentiate into hepatocytes in vivo.Nat Med. 2000; 6: 1229-1234Crossref PubMed Scopus (2130) Google Scholar, 16Schwartz RE Reyes M Koodie L Jiang Y Blackstad M Lund T Lenvik T Johnson S Hu WS Verfaillie CM Multipotent adult progenitor cells from bone marrow differentiate into functional hepatocyte-like cells.J Clin Invest. 2002; 109: 1291-1302Crossref PubMed Scopus (1103) Google Scholar It also has been shown that pancreas-derived stem cells can become liver17Wang X Al-Dhalimy M Lagasse E Finegold M Grompe M Liver repopulation and correction of metabolic liver disease by transplanted adult mouse pancreatic cells.Am J Pathol. 2001; 158: 571-579Abstract Full Text Full Text PDF PubMed Scopus (113) Google Scholar and liver stem cells can differentiate into pancreas.18Yang L Li S Hatch H Ahrens K Cornelius JG Petersen BE Peck AB In vitro trans-differentiation of adult hepatic stem cells into pancreatic endocrine hormone-producing cells.Proc Natl Acad Sci USA. 2002; 99: 8078-8083Crossref PubMed Scopus (483) Google Scholar Recently, stem/progenitor cells have been isolated from both the ducts of the exocrine pancreas,19Bonner-Weir S Taneja M Weir GC Tatarkiewicz K Song KH Sharma A O'Neil JJ In vitro cultivation of human islets from expanded ductal tissue.Proc Natl Acad Sci USA. 2000; 97: 7999-8004Crossref PubMed Scopus (905) Google Scholar, 20Ramiya VK Maraist M Arfors KE Schatz DA Peck AB Cornelius JG Reversal of insulin-dependent diabetes using islets generated in vitro from pancreatic stem cells.Nat Med. 2000; 6: 278-282Crossref PubMed Scopus (667) Google Scholar and the islets of Langerhans that make up the endocrine tissue of the pancreas.21Abraham EJ Leech CA Lin JC Zulewski H Habener JF Insulinotropic hormone glucagon-like peptide-1 differentiation of human pancreatic islet-derived progenitor cells into insulin-producing cells.Endocrinology. 2002; 143: 3152-3161Crossref PubMed Scopus (275) Google Scholar, 22Lechner A Leech CA Abraham EJ Nolan AL Habener JF Nestin-positive progenitor cells derived from adult human pancreatic islets of Langerhans contain side population (SP) cells defined by expression of the ABCG2 (BCRP1) ATP-binding cassette transporter.Biochem Biophys Res Commun. 2002; 293: 670-674Crossref PubMed Scopus (187) Google Scholar, 23Zulewski H Abraham EJ Gerlach MJ Daniel PB Moritz W Muller B Vallejo M Thomas MK Habener JF Multipotential nestin-positive stem cells isolated from adult pancreatic islets differentiate ex vivo into pancreatic endocrine, exocrine, and hepatic phenotypes.Diabetes. 2001; 50: 521-533Crossref PubMed Scopus (736) Google Scholar We have been characterizing a population of cells isolated from human pancreatic islets. These cells, nestin-positive islet-derived stem/progenitor cells (NIPs), initially express the protein nestin, a marker of neural stem cells, can be passaged extensively in vitro, and can be differentiated in vitro into islet-like clusters that produce islet hormones, eg, insulin and glucagon, by their exposure to known differentiation agents such as the insulinotropic, neogenic hormone, glucagon-like peptide-1.21Abraham EJ Leech CA Lin JC Zulewski H Habener JF Insulinotropic hormone glucagon-like peptide-1 differentiation of human pancreatic islet-derived progenitor cells into insulin-producing cells.Endocrinology. 2002; 143: 3152-3161Crossref PubMed Scopus (275) Google Scholar, 22Lechner A Leech CA Abraham EJ Nolan AL Habener JF Nestin-positive progenitor cells derived from adult human pancreatic islets of Langerhans contain side population (SP) cells defined by expression of the ABCG2 (BCRP1) ATP-binding cassette transporter.Biochem Biophys Res Commun. 2002; 293: 670-674Crossref PubMed Scopus (187) Google Scholar, 23Zulewski H Abraham EJ Gerlach MJ Daniel PB Moritz W Muller B Vallejo M Thomas MK Habener JF Multipotential nestin-positive stem cells isolated from adult pancreatic islets differentiate ex vivo into pancreatic endocrine, exocrine, and hepatic phenotypes.Diabetes. 2001; 50: 521-533Crossref PubMed Scopus (736) Google Scholar Analyses of NIPs by flow cytometry show that they contain a substantial subpopulation of side population cells, similar to pluripotent hematopoietic side population cells.22Lechner A Leech CA Abraham EJ Nolan AL Habener JF Nestin-positive progenitor cells derived from adult human pancreatic islets of Langerhans contain side population (SP) cells defined by expression of the ABCG2 (BCRP1) ATP-binding cassette transporter.Biochem Biophys Res Commun. 2002; 293: 670-674Crossref PubMed Scopus (187) Google Scholar One aspect of human NIPs that we are currently investigating is their potential efficacy to produce insulin in amounts sufficient to achieve glycemic control when transplanted into diabetic mice. Although we initiated the studies by transplanting human NIPs under the kidney capsules of immunosuppressed nude mice, we discovered that human NIPs successfully engraft when transplanted into immunocompetent C57BL/6 mice without a requirement for immunosuppression. Here we show that nestin-positive cells in the pancreas do not express either class I or class II major histocompatibility (MHC) antigens, describe the transplantation studies and some morphological characteristics of the xenografts, and demonstrate the development of mixed chimerism in the mice by the detection of donor human Y-chromosome and human-specific antigens. Sixty days after the intravenous administration of human NIPs to immunocompetent mice, we find by flow cytometry that 1.5 to 9.0% of the hematopoietic cells in spleen, bone marrow, and peripheral blood leukocytes, express human HLA class I antigens. Further, these mice given NIPs by a single systemic intravenous injection develop focal regions of chimerism in multiple organs, including intestine, kidney, pancreas, heart, skeletal muscle, and brain, as detected by in situ hybridization using a human-specific probe to repetitive ALU sequences and by human ALU-specific polymerase chain reaction (PCR). We propose that human NIPs may induce a state of immune tolerance in immunocompetent mice such that the human tissue is recognized as self by the immune system of the mice. These findings suggest that when stem/progenitor cells derived from a human adult tissue (eg, pancreatic islets) are transplanted into immunocompetent mice they appear to induce tissue tolerance, resist graft versus host disease, and differentiate into specific cellular phenotypes, defined by the expression of markers of epithelial tissue (mixed keratins) and mesenchymal tissue (vimentin) as well as the human-specific marker antigen for hematopoietic tissue [leukocyte common antigen (LCA), CD45]. We suggest that our findings may hold promise for the use of these cells in future approaches to applications for the regeneration of degenerated tissues in human diseases. Basic fibroblast growth factor and epidermal growth factor were obtained from Sigma (St. Louis, MO). Leukemia inhibitory factor was obtained from Chemicon (Temecula, CA). The Y-chromosome DNA hybridization probe labeled with Spectrum Red was purchased from Vysis Inc. (Downers Grove, IL) and antibodies were from BD Pharmingen (Lexington, KY). C57BL/6 mice, 8 to 10 weeks old, were obtained from Charles River Laboratories (Wilmington, MA) for use in the transplantation of human NIPs either under the kidney capsule or for systemic administration, by injection into the tail vein. Human islet tissue was obtained from the Juvenile Diabetes Research Foundation Center for Islet Transplantation, Harvard Medical School (Boston, MA), and the Diabetes Research Institute, University of Miami School of Medicine (Miami, FL). NIPs were isolated and propagated as described previously.23Zulewski H Abraham EJ Gerlach MJ Daniel PB Moritz W Muller B Vallejo M Thomas MK Habener JF Multipotential nestin-positive stem cells isolated from adult pancreatic islets differentiate ex vivo into pancreatic endocrine, exocrine, and hepatic phenotypes.Diabetes. 2001; 50: 521-533Crossref PubMed Scopus (736) Google Scholar Briefly, islets were washed and cultured in RPMI 1640 medium containing 10% serum, 11.1 mmol/L glucose, antibiotics, sodium pyruvate, β-mercaptoethanol, and growth factors. Within several days, nestin-positive cells grew out from islets. These cells were cloned and expanded in medium containing 20 ng/ml each of basic fibroblast growth factor and epidermal growth factor or, 10 ng/ml of human leukemia inhibitory factor (Chemicon) in the presence or absence of serum. In certain instances the cells were maintained in the absence of serum. Chromosomal analysis and karyotyping were performed at the Dana Farber/HCC cytogenetics core laboratory, Brigham and Women's Hospital, Harvard Medical School. Between 105 and 106 NIP cells prepared from human male or female donor islets were transplanted under the kidney capsules of 26 female C57BL/6 mice without immunosuppression. The transplantation procedure has been described previously.24Ryu S Kodama S Ryu K Schoenfeld DA Faustman DL Reversal of established autoimmune diabetes by restoration of endogenous beta cell function.J Clin Invest. 2001; 108: 63-72Crossref PubMed Scopus (146) Google Scholar The mice were sacrificed 15 to 60 days after the transplantations and the kidneys containing the NIP grafts were removed for histomorphological and biochemical analyses including fluorescence in situ hybridization (FISH) for the detection of human Y-chromosome, immunocytochemistry for detection of human-specific antigens, and detection of enhanced green fluorescent protein (EGFP). Four mice were each given a total of 105 to 106 NIPs intravenously via the tail vein, administered twice 1 month apart. Sixty days after the first intravenous injection of cells, the mice were sacrificed. Spleen, bone marrow, and peripheral blood were analyzed for the presence of chimerism by fluorescence-activated sorting of cells using the human marker HLA-A, -B, -C. The pancreata, kidneys, livers, hearts, skeletal muscle, intestines, brains, and lungs were collected from the mice for in situ histohybridization using a DNA probe specific for the detection of human Y-chromosome, and human repetitive ALU DNA sequences as measures of tissue microchimerism. The rabbit anti-human nestin was a gift from Dr. C. Messam (National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD). The mouse monoclonal antibody against human CD-45; HLA-A, -B, -C (class 1 HLA marker); mixed keratins; and vimentin were purchased from BD Pharmingen. MHC I and II antibodies were from Serotec Inc. (Raleigh, NC). Frozen kidneys with grafts were embedded in OCT compound and 5-μm tissue sections were prepared. Tissue sections were fixed in acetone, blocked with normal goat serum, followed by avidin D and biotin-blocking solution, and then incubated overnight in mouse anti-human antibodies to CD45 (LCA) vimentin, and mixed keratins or in anti-rat antibodies to MHC I, MHC II, as described previously.21Abraham EJ Leech CA Lin JC Zulewski H Habener JF Insulinotropic hormone glucagon-like peptide-1 differentiation of human pancreatic islet-derived progenitor cells into insulin-producing cells.Endocrinology. 2002; 143: 3152-3161Crossref PubMed Scopus (275) Google Scholar Sections were rinsed with phosphate-buffered saline (PBS) and incubated with biotinylated goat anti-mouse serum. Sections were then immersed in diaminobenzidine substrate solution and a red-brown precipitate was visualized. Immunostaining for insulin, glucagon, and PDX-1 of the subrenal capsular grafts was performed on 5-μm serial sections of frozen tissue. Antisera used were: guinea pig anti-insulin IgG and guinea pig anti-glucagon serum (Linco Research Inc., St. Charles, MO) and polyclonal antisera to human PDX-1 (IPF-1) (antiserum R25325Stoffers DA Zinkin NT Stanojevic V Clarke WL Habener JF Pancreatic agenesis attributable to a single nucleotide deletion in the human IPF1 gene coding sequence.Nat Genet. 1997; 15: 106-110Crossref PubMed Scopus (918) Google Scholar). Normal guinea pig IgG and normal rabbit serum served as controls. Immunoreactivity was developed using both the glucose oxidase method, with nitro blue tetrazolium as the substrate, and the peroxidase method, with diaminobenzidine as the substrate (Vector Laboratories, Burlingame, CA). Tissue sections were hybridized with a human ALU DNA probe kit following the recommendations of InnoGenex (San Ramon, CA). ALU sequences are repetitive DNA elements that are unique to primates. The human ALU DNA probe does not hybridize to mouse DNA at the specified hybridization conditions. Briefly, proteinase K reagent was added to cover the sections and was incubated at room temperature for 10 to 15 minutes. Sections were then washed, refixed with 1% formalin, and hybridized with the human ALU probe at 37°C overnight. Then, tissue sections were washed repeatedly with PBS-Tween 20 buffer and incubated with primary antiserum to the ALU probe followed by incubation with secondary antibody. Subsequently, sections were immersed in streptavidin-peroxidase substrate solution and the color was developed with an amino ethyl carbazole chromogen (Sigma). Sections were counterstained with hematoxylin and mounted in SuperMount for visual microscopic examination. PCR amplification of human Alu-sx and mouse c-mos sequences. DNA was extracted using the QIAamp DNA extraction kit (Qiagen, Valencia, CA). Alu-sx primers were ALU-forward: 5′-GGCGCGGTGGCTCACG-3′ and ALU-reverse: 5′-TTTTTTGAGACGGAGTCTCGCTC-3′. Primers for c-mos were MOS-forward: 5′-GAATTCAGATTTGTGCATACACAGTGACT-3′, and MOS-reverse: 5′-AACATTTTTCGGGAATAAAAGTTGAGT-3′. DNA amplification and primer selection were as previously described.26Cho JJ Malhi H Wang R Joseph B Ludlow JW Susik R Gupta S Enzymatically labeled chromosomal probes for in situ identification of human cells in xenogeneic transplant models.Nat Med. 2002; 8: 1033-1036Crossref PubMed Scopus (12) Google Scholar PCR conditions were 94°C for 5 minutes followed by 40 cycles of 94°C for 30 seconds, 58°C for 30 seconds, and 72°C for 60 seconds, and final extension 72°C for 10 minutes. PCR products were resolved by electrophoresis on a 1.5% agarose gel, and transferred overnight to a cellulose filter. Southern blot hybridization was performed by autoradiographic detection with an ALU internal probe: 5′-CCACTTTGGGAGGCCGAGGCGGGTGGATCATGAGGTACAAGGTGAAACCC-3′. Further confirmation of the presence of human sequences was achieved by cloning of the PCR fragments into the pCRII plasmid by TA cloning (Invitrogen Corp., Carlsbad, CA) and direct sequencing.26Cho JJ Malhi H Wang R Joseph B Ludlow JW Susik R Gupta S Enzymatically labeled chromosomal probes for in situ identification of human cells in xenogeneic transplant models.Nat Med. 2002; 8: 1033-1036Crossref PubMed Scopus (12) Google Scholar Human NIPs, maintained in long-term cultures containing serum and human leukemia inhibitory factor, were transfected with a plasmid expressing the EGFP according to published protocols (GenePorter; Gene Therapy Systems, Inc., San Diego, CA). Cells were transfected sequentially twice within a period of 4 days to enhance the number of transfected cells. By this method ∼20 to 30% of the cells expressed the green fluorescent protein. FISH was performed with a commercially available kit according to the manufacturer's recommendations (Vysis). Briefly, 5-μm frozen sections on silanized slides were fixed with methanol and acetic acid, washed with PBS and 2× standard saline citrate at 73°C for 2 minutes. Then, sections were immersed in pepsin solution at 37°C for 5 minutes, washed in 2× standard saline citrate at room temperature for 1 minute, and dehydrated for 1 minute in 85% and 100% ethanol successively. The CEP Y-chromosome probe (orange fluorescence) mixture was denatured at 73°C, and applied onto the sections. Hybridization was performed at 37°C. Sixteen hours later, the slides were washed in 2× standard saline citrate for 1 to 4 minutes and mounted in a 4,6-diamidino-2-phenylindole (blue fluorescence)-containing solution to allow visualization of the nuclei. Peripheral blood leukocytes and bone marrow cells as well as splenocytes were isolated for flow cytometry analysis.27Ikehara Y Yasunami Y Kodama S Maki T Nakano M Nakayama T Taniguchi M Ikeda S CD4(+) Valpha14 natural killer T cells are essential for acceptance of rat islet xenografts in mice.J Clin Invest. 2000; 105: 1761-1767Crossref PubMed Scopus (138) Google Scholar Cells were washed in Hanks' buffer containing 10% bovine serum albumin, incubated with an Fc-receptor blocking solution, and then incubated with phycoerythrin-labeled mouse anti-human HLA-A, B, C antigen. Cells obtained from animals that received tail vein injections of human NIPs, as well as control mice that did not receive NIPs, were analyzed by flow cytometry. We have described previously the procedures for the isolation and culture of the NIPs.21Abraham EJ Leech CA Lin JC Zulewski H Habener JF Insulinotropic hormone glucagon-like peptide-1 differentiation of human pancreatic islet-derived progenitor cells into insulin-producing cells.Endocrinology. 2002; 143: 3152-3161Crossref PubMed Scopus (275) Google Scholar, 22Lechner A Leech CA Abraham EJ Nolan AL Habener JF Nestin-positive progenitor cells derived from adult human pancreatic islets of Langerhans contain side population (SP) cells defined by expression of the ABCG2 (BCRP1) ATP-binding cassette transporter.Biochem Biophys Res Commun. 2002; 293: 670-674Crossref PubMed Scopus (187) Google Scholar, 23Zulewski H Abraham EJ Gerlach MJ Daniel PB Moritz W Muller B Vallejo M Thomas MK Habener JF Multipotential nestin-positive stem cells isolated from adult pancreatic islets differentiate ex vivo into pancreatic endocrine, exocrine, and hepatic phenotypes.Diabetes. 2001; 50: 521-533Crossref PubMed Scopus (736) Google Scholar We have now examined the karyotype of the NIPs. Analysis of chromosome preparations of a NIP culture (clone Hu-6a, passage 06) revealed a 46, XY diploid cell line in two cells analyzed (Figure 1). Analyses of three additional cells revealed 46 chromosomes. NIP cultures were prepared from islets obtained from several different human male and female donors provided by islet procurement and transplantation centers. Human NIPs (105 to 106 cells) were transplanted under the kidney capsules of 26 immunocompetent female C57BL/6 mice throughout the course of 2.5 years. The mice were sacrificed from 15 to 60 days after the transplantations. Eight of the mice died within 24 hours of causes related to the anesthesia and/or surgery. In 10 of 18 mice the NIPs successfully engrafted and grew into readily visible masses of tissue (Figure 2A). The NIPs transplanted into 8 of 18 mice failed to engraft. We then set out to determine whether the grafts contained human tissue (xenografts). First, we transfected human NIPs with a plasmid expressing EGFP as shown in Figure 2B. These transfected NIP cultures were then transplanted under the kidney capsule of a mouse. Fifteen days later, the kidney was removed and under UV light the graft showed intense EGFP fluorescence (Figure 2C). Later, the graft was excised, enzymatically dissociated, and cultured for an additional 5 days. Transfected NIPs present in explant cultures showed expression of EGFP (Figure 2D). Examination by hematoxylin and eosin staining of the histology of the grafts showed circumscribed tissue with considerable pleomorphism without evidence of invasion into the adjacent kidney parenchyma (Figure 3A). In various grafts, and tissue sections of individual grafts, focal regions of tissue were seen that had the appearance of glandular epithelial tissue (Figure 3, B and C). The predominant tissue type appeared to consist of mesenchymal stromal-like tissue (Figure 3, D and E). These observations indicate that human NIPs engraft and survive for at least 60 days after transplantation under the kidney capsules of immunocompetent mice and appear to represent different tissue types. It should be noted that heretofore transplantation of human tissue xenografts into immunocompetent mice results in a complete rejection of the graft after 5 to 10 days.24Ryu S Kodama S Ryu K Schoenfeld DA Faustman DL Reversal of established autoimmune diabetes by restoration of endogenous beta cell function.J Clin Invest. 2001; 108: 63-72Crossref PubMed Scopus (146) Google Scholar, 27Ikehara Y Yasunami Y Kodama S Maki T Nakano M Nakayama T Taniguchi M Ikeda S CD4(+) Valpha14 natural killer T cells are essential for acceptance of rat islet xenografts in mice.J Clin Invest. 2000; 105: 1761-1767Crossref PubMed Scopus (138) Google Scholar, 28Faustman DL Coe C Prevention of xenograft rejection by masking donor HLA class I antigens.Science. 1991; 252: 1700-1702Crossref PubMed Scopus (97) Google Scholar Therefore human NIPs may have a special property by which to induce the host (mouse) to recognize them as self and not to reject the xenograft. In this regard we show that the nestin-positive cells within rat pancreatic islets do not express either MHC I or MHC II antigens (Figure 3F). Thus NIPs, as they reside in the islets, may be immunologically naive because they appear not to express either MHC I or MHC II antigens. Examination of the xenograft tissues by immunohistochemistry using antibodies specific for human-specific LCA (CD45) a marker of hematopoietic tissues, keratin, an epithelial cell marker and vimentin, a mesenchymal marker, revealed clusters of immunopositive cells within the grafts concentrated along the border of the graft with the kidney (Figure 4). The more peripherally located tissue in the graft is probably of mouse origin, indicating a host versus graft cellular proliferative response, but not typical of a vigorous graft rejection response. The reaction is more typical of a delayed sensitivity granulomatous response with fibrosis and scarring, rather than an acute rejection with lymphocytic infiltration.Figure 4Immunohistochemical analyses of human NIP grafts in immunocompetent mice. A: Immunostaining for human-specific LCA, CD45, in a section of mouse kidney transplanted (kidney capsule) with human NIPs examined 35 days after the transplant. The brown-colored cells (shown in the figure as grayish-black) are the cells in the xenograft that express human LCA in the recipient mouse. K, Kidney; G, graft. B: LCA immunostaining of the graft shown at higher magnification. C: Keratin immunostaining. D: Vimentin immunostaining. E: Detection of pancreatic endocrine hormones insulin and glucagon, and pancreas- and duodenal-specific homeobox transcription factor, PDX-1 in serial sections of expanded grafts of human NIPs 15 days after transplantation under the kidney capsules of immunocompetent C57BL/6 mice. Islands of pancreatic endocrine-like tissue are found scattered about in the graft, which grew to ∼50% the volume of the kidney without evidence of invasion or oncogenicity. The top row shows two representative fields of the grafts. GP-IgG and normal rabbit serum are the control antisera for insulin and glucagon/PDX-1, respectively (bottom row). Immunostaining done by glucose oxidase method (shown in the figure as grayish-black). Immunocytochemistry was also done by diaminobenzidine (Vectastain ABC) with similar results. Asterisk denotes a b" @default.
- W2073293485 created "2016-06-24" @default.
- W2073293485 creator A5000148876 @default.
- W2073293485 creator A5010327980 @default.
- W2073293485 creator A5034913449 @default.
- W2073293485 creator A5051569390 @default.
- W2073293485 creator A5052388488 @default.
- W2073293485 creator A5084390948 @default.
- W2073293485 date "2004-03-01" @default.
- W2073293485 modified "2023-10-14" @default.
- W2073293485 title "Human Pancreatic Islet-Derived Progenitor Cell Engraftment in Immunocompetent Mice" @default.
- W2073293485 cites W124239641 @default.
- W2073293485 cites W1518831632 @default.
- W2073293485 cites W1542326554 @default.
- W2073293485 cites W1965998502 @default.
- W2073293485 cites W1985267792 @default.
- W2073293485 cites W1991374176 @default.
- W2073293485 cites W2000351395 @default.
- W2073293485 cites W2013568009 @default.
- W2073293485 cites W2020672759 @default.
- W2073293485 cites W2022109398 @default.
- W2073293485 cites W2030166471 @default.
- W2073293485 cites W2032042635 @default.
- W2073293485 cites W2037343561 @default.
- W2073293485 cites W2039396493 @default.
- W2073293485 cites W2048056915 @default.
- W2073293485 cites W2048258494 @default.
- W2073293485 cites W2049318896 @default.
- W2073293485 cites W2052530372 @default.
- W2073293485 cites W2054721944 @default.
- W2073293485 cites W2062813847 @default.
- W2073293485 cites W2063381533 @default.
- W2073293485 cites W2068375644 @default.
- W2073293485 cites W2069100407 @default.
- W2073293485 cites W2079733011 @default.
- W2073293485 cites W2084652820 @default.
- W2073293485 cites W2084936584 @default.
- W2073293485 cites W2098015360 @default.
- W2073293485 cites W2102777081 @default.
- W2073293485 cites W2104898886 @default.
- W2073293485 cites W2108143410 @default.
- W2073293485 cites W2109191124 @default.
- W2073293485 cites W2109794889 @default.
- W2073293485 cites W2110525047 @default.
- W2073293485 cites W2126088489 @default.
- W2073293485 cites W2143801694 @default.
- W2073293485 cites W2146383332 @default.
- W2073293485 cites W2152918507 @default.
- W2073293485 cites W2156035735 @default.
- W2073293485 cites W2156959752 @default.
- W2073293485 cites W2158048826 @default.
- W2073293485 cites W2158461247 @default.
- W2073293485 cites W2164035061 @default.
- W2073293485 cites W2172294715 @default.
- W2073293485 cites W245016890 @default.
- W2073293485 cites W4232584760 @default.
- W2073293485 doi "https://doi.org/10.1016/s0002-9440(10)63170-7" @default.
- W2073293485 hasPubMedCentralId "https://www.ncbi.nlm.nih.gov/pmc/articles/1613272" @default.
- W2073293485 hasPubMedId "https://pubmed.ncbi.nlm.nih.gov/14982836" @default.
- W2073293485 hasPublicationYear "2004" @default.
- W2073293485 type Work @default.
- W2073293485 sameAs 2073293485 @default.
- W2073293485 citedByCount "45" @default.
- W2073293485 countsByYear W20732934852012 @default.
- W2073293485 countsByYear W20732934852014 @default.
- W2073293485 countsByYear W20732934852015 @default.
- W2073293485 crossrefType "journal-article" @default.
- W2073293485 hasAuthorship W2073293485A5000148876 @default.
- W2073293485 hasAuthorship W2073293485A5010327980 @default.
- W2073293485 hasAuthorship W2073293485A5034913449 @default.
- W2073293485 hasAuthorship W2073293485A5051569390 @default.
- W2073293485 hasAuthorship W2073293485A5052388488 @default.
- W2073293485 hasAuthorship W2073293485A5084390948 @default.
- W2073293485 hasBestOaLocation W20732934851 @default.
- W2073293485 hasConcept C134018914 @default.
- W2073293485 hasConcept C142724271 @default.
- W2073293485 hasConcept C15729860 @default.
- W2073293485 hasConcept C165220095 @default.
- W2073293485 hasConcept C201750760 @default.
- W2073293485 hasConcept C203014093 @default.
- W2073293485 hasConcept C2778764654 @default.
- W2073293485 hasConcept C2779855799 @default.
- W2073293485 hasConcept C28328180 @default.
- W2073293485 hasConcept C555293320 @default.
- W2073293485 hasConcept C71924100 @default.
- W2073293485 hasConcept C86803240 @default.
- W2073293485 hasConcept C95444343 @default.
- W2073293485 hasConceptScore W2073293485C134018914 @default.
- W2073293485 hasConceptScore W2073293485C142724271 @default.
- W2073293485 hasConceptScore W2073293485C15729860 @default.
- W2073293485 hasConceptScore W2073293485C165220095 @default.
- W2073293485 hasConceptScore W2073293485C201750760 @default.
- W2073293485 hasConceptScore W2073293485C203014093 @default.
- W2073293485 hasConceptScore W2073293485C2778764654 @default.
- W2073293485 hasConceptScore W2073293485C2779855799 @default.
- W2073293485 hasConceptScore W2073293485C28328180 @default.
- W2073293485 hasConceptScore W2073293485C555293320 @default.
- W2073293485 hasConceptScore W2073293485C71924100 @default.